Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Nihon Yakurigaku Zasshi ; 159(2): 83-89, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38432924

RESUMO

Cardiotoxicity is a serious adverse effect of anti-cancer drugs. Anti-cancer drug-induced cardiotoxicity are arrhythmia, cardiac contractile dysfunction, coronary artery disease, and hypertension, which affect to the quality of life in patients with cancer. In particular, cardiac contractile dysfunction is a life-threatening symptom leading to heart failure, suggesting that it is very important to predict the risk of developing the contractile dysfunction by anti-cancer drugs. Recently, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to assess the risk of drug-induced arrhythmias. This prompts us to evaluate other cardiotoxic effects such as contractility dysfunction and structural toxicity with hiPSC-CMs. Since anti-cancer drug-induced contractility dysfunction are considered to be induced by chronic exposure, we have developed a method to assess chronic contractility dysfunction by imaging analysis of hiPSC-CMs. BMS-986094, which failed in clinical trials due to the occurrence of heart failure, was used as a positive compound. We found that chronic exposure to BMS-986094 decreased the contraction and relaxation velocity in hiPSC-CMs. Doxorubicin was observed to decrease cytotoxicity and both contraction and relaxation velocities in hiPSC-CMs. We are currently further evaluating other anti-cancer drugs with different mode-of-actions using hiPSC-CMs and assess the predictivity and utility of contractile assessment using hiPSC-CMs by comparing with real-world data. Here, we introduce our novel method to assess the chronic contractility of hiPSC-CMs by imaging analysis and discuss the future perspectives for assessing the anti-cancer drug-induced cardiotoxicity.


Assuntos
Antineoplásicos , Insuficiência Cardíaca , Células-Tronco Pluripotentes Induzidas , Humanos , Cardiotoxicidade/etiologia , Qualidade de Vida , Antineoplásicos/efeitos adversos , Insuficiência Cardíaca/induzido quimicamente , Medição de Risco
2.
Radiother Oncol ; 190: 110004, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37972738

RESUMO

PURPOSE: Despite technological advances in radiotherapy (RT), cardiotoxicity remains a common complication in patients with lung, oesophageal and breast cancers. Statin therapy has been shown to have pleiotropic properties beyond its lipid-lowering effects. Previous murine models have shown statin therapy can reduce short-term functional effects of whole-heart irradiation. In this study, we assessed the efficacy of atorvastatin in protecting against the late effects of radiation exposure on systolic function, cardiac conduction, and atrial natriuretic peptide (ANP) following a clinically relevant partial-heart radiation exposure. MATERIALS AND METHODS: Female, 12-week old, C57BL/6j mice received an image-guided 16 Gy X-ray field to the base of the heart using a small animal radiotherapy research platform (SARRP), with or without atorvastatin from 1 week prior to irradiation until the end of the experiment. The animals were followed for 50 weeks with longitudinal transthoracic echocardiography (TTE) and electrocardiography (ECG) every 10 weeks, and plasma ANP every 20 weeks. RESULTS: At 30-50 weeks, mild left ventricular systolic function impairment observed in the RT control group was less apparent in animals receiving atorvastatin. ECG analysis demonstrated prolongation of components of cardiac conduction related to the heart base at 10 and 30 weeks in the RT control group but not in animals treated with atorvastatin. In contrast to systolic function, conduction disturbances resolved at later time-points with radiation alone. ANP reductions were lower in irradiated animals receiving atorvastatin at 30 and 50 weeks. CONCLUSIONS: Atorvastatin prevents left ventricular systolic dysfunction, and the perturbation of cardiac conduction following partial heart irradiation. If confirmed in clinical studies, these data would support the use of statin therapy for cardioprotection during thoracic radiotherapy.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Disfunção Ventricular Esquerda , Humanos , Feminino , Camundongos , Animais , Cardiotoxicidade/etiologia , Cardiotoxicidade/prevenção & controle , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , Camundongos Endogâmicos C57BL , Coração/efeitos da radiação , Modelos Animais de Doenças
3.
Sci Rep ; 13(1): 22155, 2023 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-38092839

RESUMO

Cardiotoxicity limits the use of anthracyclines as potent chemotherapeutics. We employ classical molecular dynamics to explore anthracycline interactions with a realistic myocardial membrane and compare to an ideal membrane widely used in literature. The interaction of these two membranes with four anthracyclines; doxorubicin, epirubicin, daunorubicin, and idarubicin are studied. Careful analysis was conducted on three forms of each drug; pristine, primary metabolite, and cationic salt. By examining the molecular residence time near the membrane's surface, the average number of molecule/membrane hydrogen bonds, the immobilization of the molecules near the membrane, and the location of those molecules relative to the mid-plane of the membrane we found out that salt forms exhibit the highest cardiotoxic probability, followed by the metabolites and pristine forms. Additionally, all forms have more affinity to the upper layer of the realistic myocardial membrane. Meanwhile, an ideal membrane consisting of a single type of phospholipids is not capable of capturing the specific interactions of each drug form. These findings confirm that cardiotoxic mechanisms are membrane-layer and drug-form dependent.


Assuntos
Antraciclinas , Neoplasias , Humanos , Antraciclinas/efeitos adversos , Cardiotoxicidade/etiologia , Fosfolipídeos , Antibióticos Antineoplásicos/uso terapêutico , Doxorrubicina , Neoplasias/tratamento farmacológico
4.
Chem Biol Interact ; 386: 110777, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37879593

RESUMO

Cardiovascular diseases pose a huge threat to global human health and are also a major obstacle to drug development and disease treatment. Drug-induced cardiotoxicity remains an important clinical issue. Both traditional two-dimensional (2D) monolayer cell models and animal models have their own limitations and are not fully suitable for the study of human heart physiology or pathology. Cardiac organoids are three-dimensional (3D) and self-organized structures that accurately retain the biological characteristics and functions of heart tissue. In this study, we successfully established a human cardiac organoid model by inducing the directed differentiation of human embryonic stem cells, which recapitulates the patterns of early myocardial development. Moreover, this model accurately characterized the cardiotoxic damage caused by the anticancer drug doxorubicin, including clinical cardiac injury and cardiac function indicators, cell apoptosis, inflammation, fibrosis, as well as mitochondrial damage. In general, the cardiac organoid model can be used to evaluate the cardiotoxicity of drugs, opening new directions and ideas for drug screening and cardiotoxicity research.


Assuntos
Cardiotoxicidade , Coração , Animais , Humanos , Cardiotoxicidade/etiologia , Cardiotoxicidade/tratamento farmacológico , Doxorrubicina/toxicidade , Apoptose , Organoides , Miócitos Cardíacos
6.
Molecules ; 28(18)2023 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-37764363

RESUMO

The assessment of cardiotoxicity is a persistent problem in medicinal chemistry. Quantitative structure-activity relationships (QSAR) are one possible way to build up models for cardiotoxicity. Here, we describe the results obtained with the Monte Carlo technique to develop hybrid optimal descriptors correlated with cardiotoxicity. The predictive potential of the cardiotoxicity models (pIC50, Ki in nM) of piperidine derivatives obtained using this approach provided quite good determination coefficients for the external validation set, in the range of 0.90-0.94. The results were best when applying the so-called correlation intensity index, which improves the predictive potential of a model.


Assuntos
Cardiotoxicidade , Química Farmacêutica , Humanos , Cardiotoxicidade/etiologia , Método de Monte Carlo , Piperidinas , Relação Quantitativa Estrutura-Atividade
7.
Adv Sci (Weinh) ; 10(30): e2301136, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37679058

RESUMO

Doxorubicin-induced cardiomyopathy (DIC) brings tough clinical challenges as well as continued demand in developing agents for adjuvant cardioprotective therapies. Here, a zebrafish phenotypic screening with deep-learning assisted multiplex cardiac functional analysis using motion videos of larval hearts is established. Through training the model on a dataset of 2125 labeled ventricular images, ZVSegNet and HRNet exhibit superior performance over previous methods. As a result of high-content phenotypic screening, cyanidin chloride (CyCl) is identified as a potent suppressor of DIC. CyCl effectively rescues cardiac cell death and improves heart function in both in vitro and in vivo models of Doxorubicin (Dox) exposure. CyCl shows strong inhibitory effects on lipid peroxidation and mitochondrial damage and prevents ferroptosis and apoptosis-related cell death. Molecular docking and thermal shift assay further suggest a direct binding between CyCl and Keap1, which may compete for the Keap1-Nrf2 interaction, promote nuclear accumulation of Nrf2, and subsequentially transactivate Gpx4 and other antioxidant factors. Site-specific mutation of R415A in Keap1 significantly attenuates the protective effects of CyCl against Dox-induced cardiotoxicity. Taken together, the capability of deep-learning-assisted phenotypic screening in identifying promising lead compounds against DIC is exhibited, and new perspectives into drug discovery in the era of artificial intelligence are provided.


Assuntos
Cardiotoxicidade , Aprendizado Profundo , Animais , Cardiotoxicidade/etiologia , Cardiotoxicidade/prevenção & controle , Peixe-Zebra/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Inteligência Artificial , Simulação de Acoplamento Molecular , Estresse Oxidativo , Doxorrubicina/toxicidade
8.
Wiad Lek ; 76(7): 1600-1607, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37622503

RESUMO

OBJECTIVE: The aim: To evaluate the effect of Necrostatin-1s (Nec-1s), an inhibitor of necroptosis, on acute Dox-induced cardiotoxicity in a mice model. PATIENTS AND METHODS: Materials and methods: Fifteen male mice were used. The animals were allocated into three groups. On the third day of the experiment, a single intraper¬itoneal dose of 20 mg/kg Dox was used to induce cardiotoxicity. Mice in the control group were given vehicle (DMSO) intraperitoneally, whereas mice in the third group were given 5 mg/kg Nec-1s two days before Dox treatment and continued for a total of five days. Animals were euthanized at the conclusion of the research. ELISA was used to assess the following parameters: cTnI, TNF-α, IL-1ß, GPX-4, and Hmox-1. The expression of TNF-R1 and phosphorylated NF-κß p65 was measured using immunohistochemistry. In addition, a histopathologic evaluation of the cardiac lesions was conducted. RESULTS: Results: Our results showed that Dox treatment substantially elevated serum cTnI levels, increased tissue inflammatory biomarkers (TNF-α, IL-1ß, phospho NF-κß p65 and TNF-R1), and reduced tissue antioxidant enzymes (GPX-4, Hmox-1). A histopathological analysis showed pronounced necrosis and vacuolization. These results were drastically changed by pretreatment with Nec-1s, with serum cTnI levels in this group being much lower than in the Dox group. In addition to a significant decrease in inflammatory markers, antioxidant enzymes were partially recovered. Moreover, there was preservation of the cardiac morphology to a level that was roughly normal. CONCLUSION: Conclusions: Our findings demonstrate that pretreatment with Nec-1s protected against acute Dox-induced cardiotoxicity. This cardioprotective effect was mainly due to amelioration of inflammation that reflected by inhibition of NF-κß/TNF-α/TNF-R1 pathway, with partial restoration of antioxidant enzymes, GPX-4 and Hmox1.


Assuntos
Antioxidantes , Cardiotoxicidade , Masculino , Animais , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/etiologia , Cardiotoxicidade/prevenção & controle , Receptores Tipo I de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa , Modelos Animais de Doenças , Doxorrubicina/efeitos adversos
9.
Cancer Rep (Hoboken) ; 6(9): e1852, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37354068

RESUMO

BACKGROUND: The growing population of long-term childhood cancer survivors encounter a substantial burden of cardiovascular complications. The highest risk of cardiovascular complications is associated with exposure to anthracyclines and chest radiation. Longitudinal cardiovascular surveillance is recommended for childhood cancer patients; however, the optimal methods and timing are yet to be elucidated. AIMS: We aimed to investigate the feasibility of different echocardiographic methods to evaluate left ventricular systolic function in retrospective datasets, including left ventricular ejection fraction (LVEF), fractional shortening (FS), global longitudinal strain (GLS) and longitudinal strain (LS) as well as the incidence and timing of subclinical left ventricular dysfunction detected by these methods. METHODS AND RESULTS: A retrospective longitudinal study was performed with re-analysis of longitudinal echocardiographic data, acquired during treatment and early follow-up, including 41 pediatric sarcoma patients, aged 2.1-17.8 years at diagnosis, treated at Astrid Lindgren Children's Hospital, Stockholm, Sweden, during the period 2010-2021. All patients had received treatment according to protocols including high cumulative doxorubicin equivalent doses (≥250 mg/m2 ). In 68% of all 366 echocardiograms, LS analysis was feasible. Impaired LS values (<17%) was demonstrated in >40%, with concomitant impairment of either LVEF or FS in 20% and combined impairment of both LVEF and FS in <10%. Importantly, there were no cases of abnormal LVEF and FS without concomitant LS impairment. CONCLUSION: Our findings demonstrate feasibility of LS in a majority of echocardiograms and a high incidence of impaired LS during anthracycline treatment for childhood sarcoma. We propose inclusion of LS in pediatric echocardiographic surveillance protocols.


Assuntos
Antraciclinas , Sarcoma , Criança , Humanos , Antraciclinas/efeitos adversos , Cardiotoxicidade/diagnóstico , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/etiologia , Volume Sistólico , Função Ventricular Esquerda , Estudos Longitudinais , Estudos Retrospectivos , Antibióticos Antineoplásicos , Sarcoma/tratamento farmacológico
10.
Eur J Nucl Med Mol Imaging ; 50(3): 792-812, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36334105

RESUMO

Cardiotoxicity may present as (pulmonary) hypertension, acute and chronic coronary syndromes, venous thromboembolism, cardiomyopathies/heart failure, arrhythmia, valvular heart disease, peripheral arterial disease, and myocarditis. Many of these disease entities can be diagnosed by established cardiovascular diagnostic pathways. Nuclear medicine, however, has proven promising in the diagnosis of cardiomyopathies/heart failure, and peri- and myocarditis as well as arterial inflammation. This article first outlines the spectrum of cardiotoxic cancer therapies and the potential side effects. This will be complemented by the definition of cardiotoxicity using non-nuclear cardiovascular imaging (echocardiography, CMR) and biomarkers. Available nuclear imaging techniques are then presented and specific suggestions are made for their application and potential role in the diagnosis of cardiotoxicity.


Assuntos
Antineoplásicos , Cardiomiopatias , Insuficiência Cardíaca , Miocardite , Neoplasias , Medicina Nuclear , Humanos , Antineoplásicos/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Cardiotoxicidade/diagnóstico por imagem , Cardiotoxicidade/etiologia , Cardiotoxicidade/tratamento farmacológico , Miocardite/induzido quimicamente , Miocardite/tratamento farmacológico
11.
Int J Mol Sci ; 23(15)2022 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-35897819

RESUMO

Contemporary anticancer immunotherapy with chimeric antigen receptor T-cell (CAR-T) therapy has dramatically changed the treatment of many hematologic malignancies previously associated with poor prognosis. The clinical improvement and the survival benefit unveiled the risk of cardiotoxicity, ranging from minimal effects to severe cardiac adverse events, including death. Immunotherapy should also be proposed even in patients with pre-existing cardiovascular risk factors, thereby increasing the potential harm of cardiotoxicity. CAR-T therapy frequently results in cytokine release syndrome (CRS), and inflammatory activation is sustained by circulating cytokines that foster a positive feedback mechanism. Prompt diagnosis and treatment of CAR-T cardiotoxicity might significantly improve outcomes and reduce the burden associated with cardiovascular complications. Clinical and echocardiographic examinations are crucial to perform a tailored evaluation and follow-up during CAR-T treatment. This review aims to summarize the pathophysiology, clinical implications, and echocardiographic assessment of CAR-T-related cardiotoxicity to enlighten new avenues for future research.


Assuntos
Receptores de Antígenos Quiméricos , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/etiologia , Ecocardiografia , Humanos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T , Receptores de Antígenos Quiméricos/uso terapêutico , Linfócitos T
12.
ESC Heart Fail ; 9(3): 1914-1919, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35355425

RESUMO

AIMS: Tyrosine kinase inhibitors (TKIs) used to treat chronic myeloid leukaemia (CML) can cause cardiovascular adverse events. So far, the Systematic Coronary Risk Evaluation (SCORE) charts of the European Society of Cardiology (ESC) have been used to identify cancer patients at increased cardiovascular risk. The primary aim of our study was to evaluate the usefulness of the new cardiovascular risk assessment model proposed by the Cardio-Oncology Study Group of the Heart Failure Association (HFA) of the ESC in collaboration with the International Cardio-Oncology Society (ICOS) to stratify the cardiovascular risk in CML patients, compared with SCORE risk charts. The secondary aim was to establish the incidence of adverse arterial events (AEs) in patients with CML treated with TKIs and the influence of preventive treatment with aspirin. METHODS AND RESULTS: A retrospective single-centre observational study was carried out on 58 patients (32 men and 26 women; mean age ± SD: 59 ± 15 years) with CML treated with TKIs for a median period of 43 ± 31 months. Cardiological evaluation was performed and cardiovascular risk was estimated with SCORE risk charts and with the new risk assessment tool proposed by HFA/ICOS. AEs were recorded. According to SCORE charts and the new HFA/ICOS risk stratification tool, respectively, 46% (Group A1) and 60% (Group A2) of patients were at high-very high risk, and 54% (Group B1) and 40% (Group B2) at low-moderate risk. AEs were significantly more frequent in Group A1 than Group B1 (P value < 0.01) when considered overall; they were significantly more frequent in Group A2 than Group B2 either overall or considered individually. HFA/ICOS risk stratification tool was significantly more sensitive than SCORE (P < 0.01) in identifying patients at higher risk of cardiovascular toxicity. In addition, we did not find AEs in patients pretreated with aspirin. CONCLUSIONS: The new HFA/ICOS risk stratification model allows a more tailored cardiovascular risk stratification in patients with CML and it is more sensitive than SCORE charts.


Assuntos
Insuficiência Cardíaca , Leucemia Mielogênica Crônica BCR-ABL Positiva , Adulto , Idoso , Aspirina , Cardiotoxicidade/etiologia , Doença Crônica , Feminino , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/epidemiologia , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis , Leucemia Mielogênica Crônica BCR-ABL Positiva/complicações , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Medição de Risco
13.
Biomed Pharmacother ; 148: 112695, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35158142

RESUMO

Cardiotoxicity remains the most common reason for failure during drug development. Recently, the zebrafish (Danio rerio) model has emerged for the evaluation of drug-dependent cardiotoxicity and for the identification of cardioprotective molecules. However, it remains unknown how closely the zebrafish-based results may be translated to humans. To tackle this issue, we established embryonic zebrafish models of doxorubicin-, adrenaline- and terfenadine-induced cardiotoxicity with unified dosing regimen which eventually enabled head-to-head comparison of the drugs. Subsequently, we determined whether human cardioprotective medications - dexrazoxane, metoprolol, carvedilol and valsartan - are able to manage heart dysfunction in zebrafish. Our results indicated that doxorubicin, adrenaline and terfenadine elicited overt signs of cardiotoxicity in fish, and we further showed that the blockade of the renin-angiotensin system and, to a lesser extent, ß-adrenergic system, ameliorated the heart disease in zebrafish. From the drug development standpoint, our work opens the possibility to determine the cardiovascular properties of tested compounds using the rapid and affordable zebrafish model.


Assuntos
Cardiomiopatias , Cardiotoxicidade , Animais , Cardiomiopatias/induzido quimicamente , Cardiotoxicidade/etiologia , Carvedilol/farmacologia , Doxorrubicina/toxicidade , Peixe-Zebra
14.
Cancer Med ; 11(8): 1763-1768, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35166044

RESUMO

Graphical AbstractThe aim was to assess the late cardiotoxic effects in young adults treated for various cancer types in childhood using echocardiography and 24-h ECG Holter monitoring.


Assuntos
Eletrocardiografia Ambulatorial , Neoplasias , Cardiotoxicidade/etiologia , Ecocardiografia , Humanos , Neoplasias/tratamento farmacológico , Adulto Jovem
15.
Clin Lymphoma Myeloma Leuk ; 22(1): e57-e64, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34446381

RESUMO

BACKGROUND: Doxorubicin carries a risk of congestive heart failure (CHF). Black race has been suggested as a risk factor for doxorubicin-related cardiotoxicity, but data are limited. We assessed whether HF occurs at higher rates in Black patients compared to White patients who receive doxorubicin for DLBCL, and evaluated race as an independent risk factor for the development of HF after adjusting for known risk factors. PATIENTS AND METHODS: We used SEER-Medicare to identify patients 66 years and older with DLBCL. We excluded patients with CHF documented prior to diagnosis with DLBCL. We assessed for hypertension, type 2 diabetes, coronary artery disease, and arrhythmias prior to diagnosis with DLBCL. The primary outcome was documented CHF at any point following DLBCL diagnosis. Secondary outcomes included CHF in the first year following diagnosis and death. We performed analyses additionally stratified by cumulative dose of doxorubicin. RESULTS: Our study population consisted of 8,604 patients (White 96.8%, Black 3.2%). In both Kaplan-Meier and competing risk analyses, we observed no significant difference in the incidence of CHF between Black and White patients, both before and after adjusting for covariates. Finally, we observed no significant differences in the incidence of CHF by race after stratification by cumulative doxorubicin dose. CONCLUSIONS: CHF is common following doxorubicin chemotherapy for DLBCL in older patients. No association was observed between Black race and the onset of heart failure in this setting. Rigorous screening for known clinical risk factors is likely more relevant than race in treatment selection and optimization.


Assuntos
Cardiotoxicidade/etiologia , Doxorrubicina/efeitos adversos , Insuficiência Cardíaca/induzido quimicamente , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Cardiotoxicidade/fisiopatologia , Feminino , Humanos , Incidência , Linfoma Difuso de Grandes Células B/patologia , Masculino , Grupos Raciais , Estudos Retrospectivos , Medição de Risco , Fatores de Risco
16.
Eur Rev Med Pharmacol Sci ; 25(19): 6013-6024, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34661261

RESUMO

OBJECTIVE: Methotrexate (MTX) is one of the most commonly used anti-cancer drugs for various types of neoplasms. It is associated with multiple cytotoxic effects including nephrotoxicity, hepatotoxicity and cardiotoxicity. Liraglutide (LIR) is a potent anti-diabetic drug and also has antioxidant and anti-inflammatory properties. In this study, we tried to investigate the protective effect of LIR on MTX induced cardiotoxicity and to identify the molecular mechanisms for this protection. MATERIALS AND METHODS:  Rats were divided into 4 groups, including control group, LIR group, MTX group and LIR + MTX group. ECG was measured then blood samples were taken, and hearts were excised for biochemical and histological investigations. RESULTS: MTX group exhibited a mild non-significant irregular bradycardia, an increase of CK-MB besides a decrease of total antioxidant capacity. MTX administration also resulted in downregulation of vascular endothelial growth factor (VEGF), while caused upregulation of interleukin 1 beta (IL-1B) and interleukin 6 (IL-6) in comparison to the control group. Also, MTX group showed histological abnormalities besides negative VEGF and positive iNOS as detected by immunohistochemical staining compared to the control group. LIR administration could reverse these results. CONCLUSIONS: LIR prevented MTX induced cardiotoxicity through its antioxidant and anti-inflammatory properties.


Assuntos
Cardiotônicos/farmacologia , Cardiotoxicidade/prevenção & controle , Liraglutida/farmacologia , Metotrexato/toxicidade , Animais , Anti-Inflamatórios/farmacologia , Antimetabólitos Antineoplásicos/toxicidade , Antioxidantes/farmacologia , Cardiotoxicidade/etiologia , Regulação para Baixo/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Inflamação/tratamento farmacológico , Inflamação/patologia , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Ratos , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética
17.
BMC Cancer ; 21(1): 1114, 2021 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-34663256

RESUMO

BACKGROUND: Today, cancer ranks as one of the leading causes of death. Despite the large number of novel available therapies, radiotherapy (RT) remains as the most effective non-surgical method to cure cancer patients. In fact, approximately 50% of all cancer patients receive some type of RT and among these 60% receive RT-treatment with a curative intent. However, as occurs with any other oncological therapy, RT treated patients may experience toxicity side effects that range from moderate to severe. Among these, cardiotoxicity represents a significant threat for premature death. Current methods evaluate cardiotoxic damage based on volumetric changes in the Left Ventricle Ejected Fraction (LVEF). Indeed, a 10% drop in LVEF is commonly used as indicator of cardiotoxicity. More recently, a number of novel techniques have been developed that significantly improve specificity and sensitivity of heart's volumetric changes and early detection of cardiotoxicity even in asymptomatic patients. Among these, the Strain by Speckle Tracking (SST) is a technique based on echocardiographic analysis that accurately evaluates myocardial deformation during the cardiac cycle (ventricular and atrial function). Studies also suggest that Magnetic Resonance Imaging (MRI) is a high-resolution technique that enables a better visualization of acute cardiac damage. METHODOLOGY: This protocol will evaluate changes in SST and MRI in cancer patients that received thoracic RT. Concomitantly, we will assess changes in serum biomarkers of cardiac damage in these patients, including: high-sensitivity cardiac Troponin-T (hscTnT), N-Terminal pro-Brain Natriuretic Peptide (NTproBNP) and Circulating Endothelial Cells (CECs), a marker of endothelial dysfunction and vascular damage. DISCUSSION: The presented protocol is to our knowledge the first to prospectively and with a multimodal approach, study serological and image biomarkers off early cardiac damage due to radiotherapy. With a practical clinical approach we will seek early changes that could potentially be in the future be linked to clinical mayor events with consequences for cancer survivors.


Assuntos
Cardiotoxicidade/diagnóstico por imagem , Ecocardiografia/métodos , Imageamento por Ressonância Magnética/métodos , Neoplasias/radioterapia , Lesões por Radiação/diagnóstico , Neoplasias da Mama/radioterapia , Cardiotoxicidade/etiologia , Protocolos Clínicos , Células Endoteliais , Neoplasias Esofágicas/radioterapia , Feminino , Humanos , Neoplasias Pulmonares/radioterapia , Masculino , Contração Miocárdica/fisiologia , Peptídeo Natriurético Encefálico/análise , Fragmentos de Peptídeos/análise , Doses de Radiação , Volume Sistólico , Troponina T/análise , Disfunção Ventricular Esquerda
18.
J Toxicol Sci ; 46(8): 359-369, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34334557

RESUMO

Predicting drug-induced side effects in the cardiovascular system is very important because it can lead to the discontinuation of new drugs/candidates or the withdrawal of marketed drugs. Although chronic assessment of cardiac contractility is an important issue in safety pharmacology, an in vitro evaluation system has not been fully developed. We previously developed an imaging-based contractility assay system to detect acute cardiotoxicity using human iPS cell-derived cardiomyocytes (hiPSC-CMs). To extend the system to chronic toxicity assessment, we examined the effects of the anti-hepatitis C virus (HCV) drug candidate BMS-986094, a guanosine nucleotide analogue, which was withdrawn from phase 2 clinical trials because of unexpected contractility toxicities. Additionally, we examined sofosbuvir, another nucleotide analogue inhibitor of HCV that has been approved as an anti-HCV drug. Motion imaging analysis revealed the difference in cardiotoxicity between the cardiotoxic BMS-986094 and the less toxic sofosbuvir in hiPSC-CMs, with a minimum of 4 days of treatment. In addition, we found that BMS-986094-induced contractility impairment was mediated by a decrease in calcium transient. These data suggest that chronic treatment improves the predictive power for the cardiotoxicity of anti-HCV drugs. Thus, hiPSC-CMs can be a useful tool to assess drug-induced chronic cardiotoxicity in non-clinical settings.


Assuntos
Antivirais/toxicidade , Cardiotoxicidade/etiologia , Guanosina Monofosfato/análogos & derivados , Guanosina/análogos & derivados , Guanosina/toxicidade , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/efeitos dos fármacos , Antivirais/efeitos adversos , Cálcio/metabolismo , Células Cultivadas , Doença Crônica , Guanosina/efeitos adversos , Guanosina Monofosfato/efeitos adversos , Guanosina Monofosfato/toxicidade , Humanos , Imagem Molecular , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Segurança
19.
Clin Sci (Lond) ; 135(14): 1649-1668, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34283204

RESUMO

In the past two decades, treatment outcomes for a wide range of malignancies have improved remarkably due to the development of novel anti-cancer therapies, including vascular endothelial growth factor inhibitors (VEGFIs) and immune checkpoint inhibitors (ICIs). Despite their unprecedented anti-tumour effects, it is becoming increasingly clear that both types of agents are associated with specific cardiovascular toxicity, including hypertension, congestive heart failure, myocarditis and acceleration of atherosclerosis. Currently, VEGFI and ICI combination therapy is recommended for the treatment of advanced renal cell carcinoma (RCC) and has shown promising treatment efficacy in other tumour types as well. Consequently, VEGFI and ICI combination therapy will most likely become an important therapeutic strategy for various malignancies. However, this combinatory approach is expected to be accompanied by a substantial increase in cardiovascular risk, as both types of agents could act synergistically to induce cardiovascular sequelae. Therefore, a comprehensive baseline assessment and adequate monitoring by specialised cardio-oncology teams is essential in case these agents are used in combination, particularly in high-risk patients. This review summarises the mechanisms of action and treatment indications for currently registered VEGFIs and ICIs, and discusses their main vascular and cardiac toxicity. Subsequently, we provide the biological rationales for the observed promising synergistic anti-tumour effects of combined VEGFI/ICI administration. Lastly, we speculate on the increased risk for cardiovascular toxicity in case these agents are used in combination and its implications and future directions for the clinical situation.


Assuntos
Inibidores da Angiogênese/efeitos adversos , Cardiotoxicidade/etiologia , Doenças Cardiovasculares/tratamento farmacológico , Inibidores de Checkpoint Imunológico/efeitos adversos , Inibidores da Angiogênese/uso terapêutico , Fatores de Risco de Doenças Cardíacas , Humanos , Hipertensão/etiologia , Hipertensão/fisiopatologia , Inibidores de Checkpoint Imunológico/uso terapêutico
20.
Curr Cardiol Rep ; 23(6): 65, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33961140

RESUMO

PURPOSE OF REVIEW: In this review, we summarize the major known cardiac toxicities of common chemotherapeutic agents and the role of nuclear cardiac imaging for the surveillance and assessment of cancer therapeutics-related cardiac dysfunction in routine clinical practice. RECENT FINDINGS: Cardiotoxicity from chemotherapy causes a significant mortality and limits potentially life-saving treatment in cancer patients. Close monitoring of cardiac function during chemotherapy is an accepted method for reducing these adverse effects especially in patients with cancer therapeutics-related cardiac dysfunction. Nuclear imaging is a sensitive, specific, and highly reproducible modality for assessment of cardiac function. Nuclear imaging techniques including equilibrium radio nucleotide angiography, myocardial perfusion imaging, and novel experimental molecular imaging are the various objective tools available in addition to conventional echocardiography and cardiac magnetic resonance imaging in the surveillance, assessment, and follow-up of cancer therapeutics-related cardiac dysfunction.


Assuntos
Antineoplásicos , Cardiopatias , Neoplasias , Antineoplásicos/efeitos adversos , Cardiotoxicidade/diagnóstico por imagem , Cardiotoxicidade/etiologia , Ecocardiografia , Cardiopatias/induzido quimicamente , Cardiopatias/diagnóstico por imagem , Humanos , Neoplasias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA